1
|
De Bartolo A, Romeo N, Marrone A, Rago V, Granieri MC, Vommaro ML, Cupelli A, Cerra MC, Indiveri C, Ronca R, Cantile M, Sanna R, Rocca C, Angelone T. A recombinant fragment antigen-binding (Fab) of trastuzumab displays low cytotoxic profile in adult human cardiomyocytes: first evidence and the key implication of FcγRIIA receptor. Acta Pharmacol Sin 2025; 46:618-631. [PMID: 39414958 PMCID: PMC11845480 DOI: 10.1038/s41401-024-01397-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/17/2024] [Indexed: 10/18/2024]
Abstract
Fragment crystallizable gamma receptors (FcγRs) mediate various cellular responses with significant cardiovascular implications. They contribute to the anticancer activity of trastuzumab (TRZ), a recombinant humanized monoclonal antibody that interferes with human epidermal growth factor receptor 2 (HER2), thereby blocking its physiological function in cardiac cells. This is responsible for cardiac complications that hamper TRZ clinical application. In this study we investigated the involvement of FcγRs in the TRZ cardiotoxicity. We used a recombinant antigen-binding fragment (Fab) of TRZ (rFab-HER2) to examine whether the absence of the Fc region resulted in fewer cardiomyocyte toxicity while preserving TRZ's ability to inhibit HER2. When exposed to rFab-HER2, AC16 human adult ventricular cardiomyocytes were less vulnerable to damage and death, than to TRZ. Specifically, TRZ exhibited cytotoxicity at a lower concentration (150 µg/mL, corresponding to ~1 µM) compared to rFab-HER2 (250 µg/mL, corresponding to ~5 µM). Like TRZ, rFab-HER2 negatively modulated HER2 levels in cardiomyocyte (without inducing cytotoxic activity in BJ human fibroblast cells that either did not express or express very low levels of HER2) and inhibited the downstream ERK/AKT cascades. But rFab-HER2 did not alter cardiomyocyte mitochondrial dynamic balance, and affect apoptosis and inflammation, while it limited cytosolic and mitochondrial ROS indicators. On contrary, the Fc region (50-250 μg/mL) exerted direct cytotoxic action on cardiomyocytes (but not on human fibroblasts that lacked Fc receptors). TRZ (150 μg/mL) markedly upregulated the expression level of FcγRIIA (a FcγRs strongly involved in TRZ-induced antibody-dependent cellular toxicity) in cardiomyocytes, whereas the Fab fragment (150 μg/mL) had no effect. Our results demonstrate that Fc region plays an important pathogenic role in TRZ-induced cardiomyocyte toxicity. In addition, targeting FcγRIIA might contribute to the off-target effects of TRZ therapy.
Collapse
Affiliation(s)
- Anna De Bartolo
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Naomi Romeo
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Alessandro Marrone
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Vittoria Rago
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Cosenza, Italy
| | - Maria Concetta Granieri
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy
| | - Maria Luigia Vommaro
- Department of Biology, E. and E. S. (DiBEST), University of Calabria, Rende, Italy
| | - Arianna Cupelli
- Department of Biology, E. and E. S. (DiBEST), Organ and System Physiology Laboratory, University of Calabria, Cosenza, Italy
| | - Maria Carmela Cerra
- Department of Biology, E. and E. S. (DiBEST), Organ and System Physiology Laboratory, University of Calabria, Cosenza, Italy
| | - Cesare Indiveri
- Department of Biology, E. and E. S. (DiBEST), Unit of Biochemistry, Molecular Biotechnology, and Molecular Biology, University of Calabria, Cosenza, Italy
- CNR Institute of Biomembranes, Bioenergetics and Molecular Biotechnology, Bari, Italy
| | | | | | | | - Carmine Rocca
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy.
- National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| | - Tommaso Angelone
- Department of Biology, E. and E. S. (DiBEST), Cellular and Molecular Cardiovascular Physiology and Pathophysiology Laboratory, University of Calabria, Cosenza, Italy.
- National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
2
|
Guha A, Shah V, Nahle T, Singh S, Kunhiraman HH, Shehnaz F, Nain P, Makram OM, Mahmoudi M, Al-Kindi S, Madabhushi A, Shiradkar R, Daoud H. Artificial Intelligence Applications in Cardio-Oncology: A Comprehensive Review. Curr Cardiol Rep 2025; 27:56. [PMID: 39969610 DOI: 10.1007/s11886-025-02215-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/06/2025] [Indexed: 02/20/2025]
Abstract
PURPOSE OF REVIEW This review explores the role of artificial intelligence (AI) in cardio-oncology, focusing on its latest application across problems in diagnosis, prognosis, risk stratification, and management of cardiovascular (CV) complications in cancer patients. It also highlights multi-omics analysis, explainable AI, and real-time decision-making, while addressing challenges like data heterogeneity and ethical concerns. RECENT FINDINGS AI can advance cardio-oncology by leveraging imaging, electronic health records (EHRs), electrocardiograms (ECG), and multi-omics data for early cardiotoxicity detection, stratification and long-term risk prediction. Novel AI-ECG models and imaging techniques improve diagnostic accuracy, while multi-omics analysis identifies biomarkers for personalized treatment. However, significant barriers, including data heterogeneity, lack of transparency, and regulatory challenges, hinder widespread adoption. AI significantly enhances early detection and intervention in cardio-oncology. Future efforts should address the impact of AI technologies on clinical outcomes, and ethical challenges, to enable broader clinical adoption and improve patient care.
Collapse
Affiliation(s)
- Avirup Guha
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Viraj Shah
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Tarek Nahle
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Shivam Singh
- Department of Internal Medicine, Reading Hospital, Tower Health, West Reading, PA, USA
| | - Harikrishnan Hyma Kunhiraman
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Fathima Shehnaz
- Department of Internal Medicine, Trinity Health Oakland, Wayne State University, Pontiac, MI, USA
| | - Priyanshu Nain
- Department of Internal Medicine, Advent Health, Rome, GA, USA
| | - Omar M Makram
- Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health Program, Michigan State University, East Lansing, MI, USA
| | - Sadeer Al-Kindi
- Division of Cardiovascular Prevention and Wellness, Houston Methodist DeBakey Heart and Vascular Center, Houston, TX, USA
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Rakesh Shiradkar
- Department of Biomedical Engineering and Informatics, Indiana University, Indianapolis, IN, USA
| | - Hisham Daoud
- School of Computer and Cyber Sciences, Augusta University, Augusta, GA, USA
| |
Collapse
|
3
|
Marimon X, Esquinas F, Ferrer M, Cerrolaza M, Portela A, Benítez R. A Novel non-invasive optical framework for simultaneous analysis of contractility and calcium in single-cell cardiomyocytes. J Mech Behav Biomed Mater 2025; 161:106812. [PMID: 39566161 DOI: 10.1016/j.jmbbm.2024.106812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 10/13/2024] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
The use of a video method based on the Digital Image Correlation (DIC) algorithm from experimental mechanics to estimate the displacements, strain field, and sarcolemma length in a beating single-cell cardiomyocyte is proposed in this work. The obtained deformation is then correlated with the calcium signal, from calcium imaging where fluorescent dyes sensitive to calcium Ca2+ are used. Our proposed video-based method for simultaneous contraction and intracellular calcium analysis results in a low-cost, non-invasive, and label-free method. This technique has shown great advantages in long-term observations because this type of intervention-free measurement neutralizes the possible alteration in the beating cardiomyocyte introduced by other techniques for measuring cell contractility (e.g., Traction Force Microscopy, Atomic Force Microscopy, Microfabrication or Optical tweezers). Three tests were performed with synthetically augmented data from cardiomyocyte images to validate the robustness of the algorithm. First, a simulated rigid translation of a referenced image is applied, then a rotation, and finally a controlled longitudinal deformation of the referenced image, thus simulating a native realistic deformation. Finally, the proposed framework is evaluated with real experimental data. To validate contraction induced by intracellular calcium concentration, this signal is correlated with a new deformation measure proposed in this article, which is independent of cell orientation in the imaging setup. Finally, based on the displacements obtained by the DIC algorithm, the change in sarcolemma length in a contracting cardiomyocyte is calculated and its temporal correlation with the calcium signal is obtained.
Collapse
Affiliation(s)
- Xavier Marimon
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain; Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain.
| | - Ferran Esquinas
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miquel Ferrer
- Department of Strength of Materials and Structural Engineering, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain
| | - Miguel Cerrolaza
- School of Engineering, Science and Technology, Valencian International University (VIU), Valencia, Spain
| | - Alejandro Portela
- Bioengineering Institute of Technology, Universitat Internacional de Catalunya (UIC), Barcelona, Spain
| | - Raúl Benítez
- Automatic Control Department, Universitat Politècnica de Catalunya (UPC-BarcelonaTECH), Barcelona, Spain; Institut de Recerca Sant Joan de Déu (IRSJD), Spain
| |
Collapse
|
4
|
Kreidieh F, McQuade J. Novel insights into cardiovascular toxicity of cancer targeted and immune therapies: Beyond ischemia with non-obstructive coronary arteries (INOCA). AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 40:100374. [PMID: 38510501 PMCID: PMC10946000 DOI: 10.1016/j.ahjo.2024.100374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Novel immune and targeted therapies approved over the past 2 decades have resulted in dramatic improvements in cancer-specific outcomes for many cancer patients. However, many of these agents can induce cardiovascular toxicity in a subset of patients. The field of cardio-oncology was established based on observations that anti-neoplastic chemotherapies and mantle radiation can lead to premature cardiomyopathy in cancer survivors. While conventional chemotherapy, targeted therapy, and immune therapies can all result in cardiovascular adverse events, the mechanisms, timing, and incidence of these events are inherently different. Many of these effects converge upon the coronary microvasculature to involve, through endocardial endothelial cells, a more direct effect through close proximity to cardiomyocyte with cellular communication and signaling pathways. In this review, we will provide an overview of emerging paradigms in the field of Cardio-Oncology, particularly the role of the coronary microvasculature in mediating cardiovascular toxicity of important cancer targeted and immune therapies. As the number of cancer patients treated with novel immune and targeted therapies grows exponentially and subsequently the number of long-term cancer survivors dramatically increases, it is critical that cardiologists and cardiology researchers recognize the unique potential cardiovascular toxicities of these agents.
Collapse
Affiliation(s)
- Firas Kreidieh
- Instructor of Clinical Medicine- Division of Hematology-Oncology; Associate Director- Internal Medicine Residency Program, American University of Beirut, Beirut, Lebanon
| | - Jennifer McQuade
- Associate Professor and Physician Scientist in Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
5
|
Attanasio U, Di Sarro E, Tricarico L, Di Lisi D, Armentaro G, Miceli S, Fioretti F, Deidda M, Correale M, Novo G, Sciacqua A, Nodari S, Cadeddu C, Tocchetti CG, Palazzuoli A, Mercurio V. Cardiovascular Biomarkers in Cardio-Oncology: Antineoplastic Drug Cardiotoxicity and Beyond. Biomolecules 2024; 14:199. [PMID: 38397436 PMCID: PMC10887095 DOI: 10.3390/biom14020199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2023] [Revised: 01/29/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Serum biomarkers represent a reproducible, sensitive, minimally invasive and inexpensive method to explore possible adverse cardiovascular effects of antineoplastic treatments. They are useful tools in risk stratification, the early detection of cardiotoxicity and the follow-up and prognostic assessment of cancer patients. In this literature review, we aim at describing the current state of knowledge on the meaning and the usefulness of cardiovascular biomarkers in patients with cancer; analyzing the intricate relationship between cancer and cardiovascular disease (especially HF) and how this affects cardiovascular and tumor biomarkers; exploring the role of cardiovascular biomarkers in the risk stratification and in the identification of chemotherapy-induced cardiotoxicity; and providing a summary of the novel potential biomarkers in this clinical setting.
Collapse
Affiliation(s)
- Umberto Attanasio
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
| | - Elena Di Sarro
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
| | - Lucia Tricarico
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (L.T.); (M.C.)
| | - Daniela Di Lisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (D.D.L.); (G.N.)
- Division of Cardiology, University Hospital Paolo Giaccone, Via del Vespro 129, 90127 Palermo, Italy
| | - Giuseppe Armentaro
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Sofia Miceli
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Francesco Fioretti
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy; (F.F.); (S.N.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy (C.C.)
| | - Michele Correale
- Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy; (L.T.); (M.C.)
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy; (D.D.L.); (G.N.)
- Division of Cardiology, University Hospital Paolo Giaccone, Via del Vespro 129, 90127 Palermo, Italy
| | - Angela Sciacqua
- Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy; (G.A.); (S.M.); (A.S.)
| | - Savina Nodari
- Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy; (F.F.); (S.N.)
| | - Christian Cadeddu
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy (C.C.)
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Center for Basic and Clinical Immunology Research (CISI), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
| | - Alberto Palazzuoli
- Cardiovascular Diseases Unit, Cardio-thoracic and Vascular Department Le Scotte Hospital, University of Siena, Strada delle Scotte 14, 53100 Siena, Italy;
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy (E.D.S.); (C.G.T.)
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
- Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
| |
Collapse
|
6
|
Langier Goncalves I, Awwad L, Aviram S, Izraeli T, Achlaug L, Aronheim A. Heart Failure Promotes Cancer Progression in an Integrin β1-Dependent Manner. Int J Mol Sci 2023; 24:17367. [PMID: 38139195 PMCID: PMC10744241 DOI: 10.3390/ijms242417367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
Heart failure and cancer are currently the deadliest diseases in the Western world, posing the most pressing clinical challenges that remain unmet today. Both conditions share similar risk factors, including age, genetics, lifestyle, chronic inflammation, stress, and more. Furthermore, medications that are being used to counteract cancer frequently result in cardiotoxicity and the spontaneous emergence of heart failure. Thus, heart failure and cancer display an intimate connection and share similarities. Recent studies show that cardiac remodeling and heart failure promote cancer progression and metastasis. Using three different mouse models for heart failure revealed that the communication between the remodeled heart and the tumor is facilitated through multiple secreted factors. Among these factors, Periostin was consistently found to be elevated in all models and was shown to be required in vitro. Yet, whether Periostin is necessary for tumor promotion in vivo is unknown. Towards this end, we examined tumor promotion in mice lacking Periostin following transverse aortic constriction (TAC). Despite the loss of Periostin, tumor growth was promoted in the TAC-operated mice. This likely occurred due to increased levels of various cytokines and growth factors in Periostin KO mice. Many of these factors are potential ligands of Integrin receptors. Therefore, we next studied the role of Integrin receptors in the tumor-promotion phenotype following heart failure. We generated cancer cells with an Integrin β1 loss of function mutation and examined tumor growth in the presence and absence of heart failure. Integrin β1 KO cancer cells fail to display cardiac-remodeling-dependent tumor-promotion. Interestingly, a previous study showed that renal cell carcinoma cells (Renca) fail to be promoted following a myocardial infarction. Consistently, we show that Renca cells do not respond to secreted factors derived from the failing heart both in vitro and in vivo. Interestingly, Renca cells display low basal mRNA levels of Integrin β1 which may explain the inability of heart failure to promote their growth. The findings may have significant clinical relevance to cardio-oncology patients who suffer from cancers with high levels of Integrin β1. Chemotherapy leading to cardiotoxicity in these patients may generate a vicious cycle with poor prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | - Ami Aronheim
- Department of Cell Biology and Cancer Science, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa 3109601, Israel; (I.L.G.); (L.A.); (S.A.); (T.I.); (L.A.)
| |
Collapse
|
7
|
Geraldes V, Caldeira E, Afonso A, Machado F, Amaro-Leal Â, Laranjo S, Rocha I. Cardiovascular Dysautonomia in Patients with Breast Cancer. Open Cardiovasc Med J 2022. [DOI: 10.2174/18741924-v16-e2206271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Breast cancer is the most frequent malignant disease among women, being responsible for a considerable percentage of fatalities and comorbidities every year. Despite advances in early detection and therapy, evidence shows that breast cancer survivors are at increased risk of developing other chronic conditions, such as cardiovascular diseases.
Autonomic dysfunction is an emerging, but poorly understood topic that has been suggested as a risk factor for cardiovascular disease in breast cancer patients. It clinically manifests through persistently elevated heart rates and abnormal heart rate variability, even before any signs of cardiovascular dysfunction appear. Since changes in the left ventricular ejection fraction only manifest when myocardial injury has already occurred, it has been hypothesized that autonomic dysfunction can constitute an early biomarker of cardiovascular impairment in breast cancer patients.
This review focuses on the direct and indirect effects of cancer and its treatment on the autonomic nervous system in breast cancer patients. We highlight the mechanisms potentially involved in cancer and antineoplastic therapy-related autonomic imbalance and review the potential strategies to prevent and/or attenuate autonomic dysfunction.
There are gaps in the current knowledge; more research in this area is needed to identify the relevance of autonomic dysfunction and define beneficial interventions to prevent cardiovascular disease in breast cancer patients.
Collapse
|
8
|
Kirichenko YY, Ilgisonis IS, Nakhodnova ES, Sokolova IY, Bochkarnikova OV, Kardanova SA, Lyapidevskaya OV, Privalova EV, Ershov VI, Belenkov YN. Case Report: AL Amyloidosis Severe Restrictive Cardiomyopathy Associated With Multiple Myeloma—Diagnostic Difficulties. Front Cardiovasc Med 2022; 9:862409. [PMID: 35770217 PMCID: PMC9235537 DOI: 10.3389/fcvm.2022.862409] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 05/05/2022] [Indexed: 11/29/2022] Open
Abstract
Background Cardiac AL amyloidosis as a complication of multiple myeloma (MM) is a formidable life-threatening condition. The first-line therapy for both MM and systemic AL amyloidosis is proteasome inhibitors (PIs). Unfortunately, the use of PIs may lead to cardiovascular toxicity development, which requires specific cardio-oncology supervision. Case Report A 57-year-old woman was admitted to a university hospital with clinical manifestation of progressive chronic heart failure. The patient had hypertension and no history of diabetes mellitus, myocardial infarction (MI), stroke, and arrhythmias. After a series of laboratory and instrumental examination methods, MM complicated by cardiac AL amyloidosis was proved. Upon specific cardio-oncology examination (NT-proBNP 4,274 pg/ml), ECHO showed systolic dysfunction, motion abnormalities in LV basal and middle segments, and a typical depositional myocardium pattern (“luminescence”); cardiac MRI revealed restrictive cardiomyopathy and specific hyperenhancement of the ventricles and atria; 24-h ECG showed QS-pattern in leads V1–V3 and unstable ventricular tachycardia (VT) paroxysms. Cardio-oncology consultation showed baseline cardiovascular risk was very high (≥20%), and cardioprotective therapy [iACE/ARBs, beta-blockers (BB), statins] was administered. The patient underwent VCD (bortezomib; cyclophosphamide; dexamethasone) chemotherapy (CMT) program. By the time of publication, the patient had received four CMT courses with a positive oncohematological and cardiovascular effect. Conclusion In this clinical case, we described a complication of MM, which was rare according to the severity and manifestation with restrictive cardiomyopathy due to secondary cardiac amyloidosis. The case's features were difficulties in verifying the underlying disease and its own complication, and the complexity of patient management according to modern principles of cardio-oncology.
Collapse
Affiliation(s)
- Yulia Y. Kirichenko
- Department of Hospital Therapy No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
- *Correspondence: Yulia Y. Kirichenko
| | - Irina S. Ilgisonis
- Department of Hospital Therapy No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Elena S. Nakhodnova
- Department of Hematology, University Hospital No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Irina Y. Sokolova
- Department of Hematology, University Hospital No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Olga V. Bochkarnikova
- Department of Hematology, University Hospital No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Sabina A. Kardanova
- Department of Hospital Therapy No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Olga V. Lyapidevskaya
- Department of Hospital Therapy No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Elena V. Privalova
- Department of Hospital Therapy No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Vladimir I. Ershov
- Department of Hospital Therapy No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| | - Yurii N. Belenkov
- Department of Hospital Therapy No. 1 of the Federal State Autonomous Educational Institution of Higher Education I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
| |
Collapse
|
9
|
Bergler-Klein J, Rainer PP, Wallner M, Zaruba MM, Dörler J, Böhmer A, Buchacher T, Frey M, Adlbrecht C, Bartsch R, Gyöngyösi M, Fürst UM. Cardio-oncology in Austria: cardiotoxicity and surveillance of anti-cancer therapies : Position paper of the Heart Failure Working Group of the Austrian Society of Cardiology. Wien Klin Wochenschr 2022; 134:654-674. [PMID: 35507087 PMCID: PMC9065248 DOI: 10.1007/s00508-022-02031-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 03/18/2022] [Indexed: 02/07/2023]
Abstract
Survival in cancer is continuously improving due to evolving oncological treatment. Therefore, cardiovascular short-term and long-term side effects gain crucial importance for overall outcome. Cardiotoxicity not only presents as heart failure, but also as treatment-resistant hypertension, acute coronary ischemia with plaque rupture or vasospasm, thromboembolism, arrhythmia, pulmonary hypertension, diastolic dysfunction, acute myocarditis and others. Recent recommendations have proposed baseline cardiac risk assessment and surveillance strategies. Major challenges are the availability of monitoring and imaging resources, including echocardiography with speckle tracking longitudinal strain (GLS), serum biomarkers such as natriuretic peptides (NT-proBNP) and highly sensitive cardiac troponins. This Austrian consensus encompasses cardiotoxicity occurrence in frequent antiproliferative cancer drugs, radiotherapy, immune checkpoint inhibitors and cardiac follow-up considerations in cancer survivors in the context of the Austrian healthcare setting. It is important to optimize cardiovascular risk factors and pre-existing cardiac diseases without delaying oncological treatment. If left ventricular ejection fraction (LVEF) deteriorates during cancer treatment (from >10% to <50%), or myocardial strain decreases (>15% change in GLS), early initiation of cardioprotective therapies (angiotensin-converting enzyme inhibitors, angiotensin or beta receptor blockers) is recommended, and LVEF should be reassessed before discontinuation. Lower LVEF cut-offs were recently shown to be feasible in breast cancer patients to enable optimal anticancer treatment. Interdisciplinary cardio-oncology cooperation is pivotal for optimal management of cancer patients.
Collapse
Affiliation(s)
- Jutta Bergler-Klein
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Peter P Rainer
- Division of Cardiology, Medical University of Graz, Graz, Austria.,BioTechMed Graz, Graz, Austria
| | - Markus Wallner
- Division of Cardiology, Medical University of Graz, Graz, Austria.,Cardiovascular Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Marc-Michael Zaruba
- Department of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Jakob Dörler
- Department of Internal Medicine III, Cardiology and Angiology, Medical University of Innsbruck, Innsbruck, Austria.,Department of Internal Medicine and Cardiology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Armin Böhmer
- Department of Internal Medicine 1, Krems University Clinic, Krems, Austria
| | - Tamara Buchacher
- Department of Internal Medicine and Cardiology, Klinikum Klagenfurt, Klagenfurt, Austria
| | - Maria Frey
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | | | - Rupert Bartsch
- Department of Medicine 1, Division of Oncology, Medical University of Vienna, Vienna, Austria
| | - Mariann Gyöngyösi
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Ursula-Maria Fürst
- Department of Internal Medicine, Hospital of the Brothers of St. John of God (Krankenhaus Barmherzige Brüder) Salzburg, Salzburg, Austria
| |
Collapse
|
10
|
Adhikari A, Asdaq SMB, Al Hawaj MA, Chakraborty M, Thapa G, Bhuyan NR, Imran M, Alshammari MK, Alshehri MM, Harshan AA, Alanazi A, Alhazmi BD, Sreeharsha N. Anticancer Drug-Induced Cardiotoxicity: Insights and Pharmacogenetics. Pharmaceuticals (Basel) 2021; 14:ph14100970. [PMID: 34681194 PMCID: PMC8539940 DOI: 10.3390/ph14100970] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
The advancement in therapy has provided a dramatic improvement in the rate of recovery among cancer patients. However, this improved survival is also associated with enhanced risks for cardiovascular manifestations, including hypertension, arrhythmias, and heart failure. The cardiotoxicity induced by chemotherapy is a life-threatening consequence that restricts the use of several chemotherapy drugs in clinical practice. This article addresses the prevalence of cardiotoxicity mediated by commonly used chemotherapeutic and immunotherapeutic agents. The role of susceptible genes and radiation therapy in the occurrence of cardiotoxicity is also reviewed. This review also emphasizes the protective role of antioxidants and future perspectives in anticancer drug-induced cardiotoxicities.
Collapse
Affiliation(s)
- Archana Adhikari
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Syed Mohammed Basheeruddin Asdaq
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Dariyah, Riyadh 13713, Saudi Arabia
- Correspondence: (S.M.B.A.); (M.C.)
| | - Maitham A. Al Hawaj
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Hofuf 31982, Saudi Arabia;
| | - Manodeep Chakraborty
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
- Correspondence: (S.M.B.A.); (M.C.)
| | - Gayatri Thapa
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Nihar Ranjan Bhuyan
- Department of Pharmaceutical Analysis, Himalayan Pharmacy Institute, Majhitar, Rangpo 737136, Sikkim, India;
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia;
| | | | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Aishah Ali Harshan
- Department of Pharmaceutical Care, Northern Area Armed Forces Hospital, King Khalid Military City Hospital, Hafr Al-Batin 39745, Saudi Arabia;
| | - Abeer Alanazi
- Department of Pharmaceutical Care, First Health Cluster in Eastern Province, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia;
| | | | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa-31982, Saudi Arabia;
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bengaluru 560035, Karnataka, India
| |
Collapse
|
11
|
Montisci A, Vietri MT, Palmieri V, Sala S, Donatelli F, Napoli C. Cardiac Toxicity Associated with Cancer Immunotherapy and Biological Drugs. Cancers (Basel) 2021; 13:4797. [PMID: 34638281 PMCID: PMC8508330 DOI: 10.3390/cancers13194797] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 12/19/2022] Open
Abstract
Cancer immunotherapy significantly contributed to an improvement in the prognosis of cancer patients. Immunotherapy, including human epidermal growth factor receptor 2 (HER2)-targeted therapies, immune checkpoint inhibitors (ICI), and chimeric antigen receptor-modified T (CAR-T), share the characteristic to exploit the capabilities of the immune system to kill cancerous cells. Trastuzumab is a monoclonal antibody against HER2 that prevents HER2-mediated signaling; it is administered mainly in HER2-positive cancers, such as breast, colorectal, biliary tract, and non-small-cell lung cancers. Immune checkpoint inhibitors (ICI) inhibit the binding of CTLA-4 or PD-1 to PDL-1, allowing T cells to kill cancerous cells. ICI can be used in melanomas, non-small-cell lung cancer, urothelial, and head and neck cancer. There are two main types of T-cell transfer therapy: tumor-infiltrating lymphocytes (or TIL) therapy and chimeric antigen receptor-modified T (CAR-T) cell therapy, mainly applied for B-cell lymphoma and leukemia and mantle-cell lymphoma. HER2-targeted therapies, mainly trastuzumab, are associated with left ventricular dysfunction, usually reversible and rarely life-threatening. PD/PDL-1 inhibitors can cause myocarditis, rare but potentially fulminant and associated with a high fatality rate. CAR-T therapy is associated with several cardiac toxic effects, mainly in the context of a systemic adverse effect, the cytokines release syndrome.
Collapse
Affiliation(s)
- Andrea Montisci
- Division of Cardiothoracic Intensive Care, ASST Spedali Civili, 25123 Brescia, Italy;
| | - Maria Teresa Vietri
- Department of Precision Medicine, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy;
| | - Vittorio Palmieri
- Department of Cardiac Surgery and Transplantation, Ospedali dei Colli Monaldi-Cotugno-CTO, 80131 Naples, Italy;
| | - Silvia Sala
- Department of Anesthesia and Intensive Care, University of Brescia, 25121 Brescia, Italy;
| | - Francesco Donatelli
- Cardiac Surgery, University of Milan, 20122 Milan, Italy
- Department of Cardiac Surgery, Istituto Clinico Sant’Ambrogio, 20149 Milan, Italy
| | - Claudio Napoli
- Clinical Department of Internal Medicine and Specialistics, University Department of Advanced Clinical and Surgical Sciences, University of Campania “Luigi Vanvitelli”, 81100 Naples, Italy;
- IRCCS SDN, 80143 Naples, Italy
| |
Collapse
|
12
|
Shraim BA, Moursi MO, Benter IF, Habib AM, Akhtar S. The Role of Epidermal Growth Factor Receptor Family of Receptor Tyrosine Kinases in Mediating Diabetes-Induced Cardiovascular Complications. Front Pharmacol 2021; 12:701390. [PMID: 34408653 PMCID: PMC8365470 DOI: 10.3389/fphar.2021.701390] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 07/14/2021] [Indexed: 12/15/2022] Open
Abstract
Diabetes mellitus is a major debilitating disease whose global incidence is progressively increasing with currently over 463 million adult sufferers and this figure will likely reach over 700 million by the year 2045. It is the complications of diabetes such as cardiovascular, renal, neuronal and ocular dysfunction that lead to increased patient morbidity and mortality. Of these, cardiovascular complications that can result in stroke and cardiomyopathies are 2- to 5-fold more likely in diabetes but the underlying mechanisms involved in their development are not fully understood. Emerging research suggests that members of the Epidermal Growth Factor Receptor (EGFR/ErbB/HER) family of tyrosine kinases can have a dual role in that they are beneficially required for normal development and physiological functioning of the cardiovascular system (CVS) as well as in salvage pathways following acute cardiac ischemia/reperfusion injury but their chronic dysregulation may also be intricately involved in mediating diabetes-induced cardiovascular pathologies. Here we review the evidence for EGFR/ErbB/HER receptors in mediating these dual roles in the CVS and also discuss their potential interplay with the Renin-Angiotensin-Aldosterone System heptapeptide, Angiotensin-(1-7), as well the arachidonic acid metabolite, 20-HETE (20-hydroxy-5, 8, 11, 14-eicosatetraenoic acid). A greater understanding of the multi-faceted roles of EGFR/ErbB/HER family of tyrosine kinases and their interplay with other key modulators of cardiovascular function could facilitate the development of novel therapeutic strategies for treating diabetes-induced cardiovascular complications.
Collapse
Affiliation(s)
- Bara A Shraim
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Moaz O Moursi
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Ibrahim F Benter
- Faculty of Medicine, Eastern Mediterranean University, Famagusta, North Cyprus
| | - Abdella M Habib
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Saghir Akhtar
- College of Medicine, QU Health, Qatar University, Doha, Qatar.,Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|
13
|
Bikiewicz A, Banach M, von Haehling S, Maciejewski M, Bielecka‐Dabrowa A. Adjuvant breast cancer treatments cardiotoxicity and modern methods of detection and prevention of cardiac complications. ESC Heart Fail 2021; 8:2397-2418. [PMID: 33955207 PMCID: PMC8318493 DOI: 10.1002/ehf2.13365] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/29/2021] [Accepted: 03/31/2021] [Indexed: 12/24/2022] Open
Abstract
The most common cancer diagnosis in female population is breast cancer, which affects every year about 2.0 million women worldwide. In recent years, significant progress has been made in oncological therapy, in systemic treatment, and in radiotherapy of breast cancer. Unfortunately, the improvement in the effectiveness of oncological treatment and prolonging patients' life span is associated with more frequent occurrence of organ complications, which are side effects of this treatment. Current recommendations suggest a periodic monitoring of the cardiovascular system in course of oncological treatment. The monitoring includes the assessment of occurrence of risk factors for cardiovascular diseases in combination with the evaluation of the left ventricular systolic function using echocardiography and electrocardiography as well as with the analysis of the concentration of cardiac biomarkers. The aim of this review was critical assessment of the breast cancer therapy cardiotoxicity and the analysis of methods its detections. The new cardio-specific biomarkers in serum, the development of modern imaging techniques (Global Longitudinal Strain and Three-Dimensional Left Ventricular Ejection Fraction) and genotyping, and especially their combined use, may become a useful tool for identifying patients at risk of developing cardiotoxicity, who require further cardiovascular monitoring or cardioprotective therapy.
Collapse
Affiliation(s)
- Agata Bikiewicz
- Heart Failure Unit, Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)Rzgowska 281/289Lodz93‐338Poland
- Department of Hypertension, Chair of Nephrology and HypertensionMedical University of LodzLodzPoland
| | - Maciej Banach
- Heart Failure Unit, Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)Rzgowska 281/289Lodz93‐338Poland
- Department of Hypertension, Chair of Nephrology and HypertensionMedical University of LodzLodzPoland
| | - Stephan von Haehling
- Department of Cardiology and Pneumology and German Center for Cardiovascular Research (DZHK), partner site GöttingenUniversity Medical Center Göttingen (UMG)GöttingenGermany
| | - Marek Maciejewski
- Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)LodzPoland
| | - Agata Bielecka‐Dabrowa
- Heart Failure Unit, Department of Cardiology and Congenital Diseases of AdultsPolish Mother's Memorial Hospital Research Institute (PMMHRI)Rzgowska 281/289Lodz93‐338Poland
- Department of Hypertension, Chair of Nephrology and HypertensionMedical University of LodzLodzPoland
| |
Collapse
|
14
|
Kobza C. Cardiac Toxicity: Using Angiotensin-Converting Enzyme Inhibitors to Prevent Anthracycline-Induced Left Ventricular Dysfunction and Cardiomyopathy. Clin J Oncol Nurs 2021; 25:259-266. [PMID: 34019025 DOI: 10.1188/21.cjon.259-266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
BACKGROUND Anthracycline chemotherapies are effective in many different types of cancer. However, cumulative doses are associated with irreversible cardiac toxicity, most frequently manifested in the development of left ventricular dysfunction, cardiomyopathy, and congestive heart failure. The onset of cardiomyopathy and subsequent heart failure can result in the interruption or discontinuation of therapy. Cardioprotective agents, particularly angiotensin- converting enzyme inhibitors, have been shown to slow the progression of left ventricular dysfunction and prevent heart failure. OBJECTIVES This review assesses the efficacy of angiotensin-converting enzyme inhibitors in the prevention of anthracycline-induced left ventricular dysfunction. METHODS A literature search was performed using four electronic databases. FINDINGS Evidence from this review suggests that angiotensin-converting enzyme inhibitors may be effective in preventing or reducing anthracycline- induced left ventricular dysfunction and subsequent cardiomyopathy and heart failure.
Collapse
|
15
|
Michalkova R, Mirossay L, Gazdova M, Kello M, Mojzis J. Molecular Mechanisms of Antiproliferative Effects of Natural Chalcones. Cancers (Basel) 2021; 13:cancers13112730. [PMID: 34073042 PMCID: PMC8198114 DOI: 10.3390/cancers13112730] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/20/2021] [Accepted: 05/27/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Despite the important progress in cancer treatment in the past decades, the mortality rates in some types of cancer have not significantly decreased. Therefore, the search for novel anticancer drugs has become a topic of great interest. Chalcones, precursors of flavonoid synthesis in plants, have been documented as natural compounds with pleiotropic biological effects including antiproliferative/anticancer activity. This article focuses on the knowledge on molecular mechanisms of antiproliferative action of chalcones and draws attention to this group of natural compounds that may be of importance in the treatment of cancer disease. Abstract Although great progress has been made in the treatment of cancer, the search for new promising molecules with antitumor activity is still one of the greatest challenges in the fight against cancer due to the increasing number of new cases each year. Chalcones (1,3-diphenyl-2-propen-1-one), the precursors of flavonoid synthesis in higher plants, possess a wide spectrum of biological activities including antimicrobial, anti-inflammatory, antioxidant, and anticancer. A plethora of molecular mechanisms of action have been documented, including induction of apoptosis, autophagy, or other types of cell death, cell cycle changes, and modulation of several signaling pathways associated with cell survival or death. In addition, blockade of several steps of angiogenesis and proteasome inhibition has also been documented. This review summarizes the basic molecular mechanisms related to the antiproliferative effects of chalcones, focusing on research articles from the years January 2015–February 2021.
Collapse
|
16
|
Green Tea ( Camellia sinensis) Extract Increased Topoisomerase II β, Improved Antioxidant Defense, and Attenuated Cardiac Remodeling in an Acute Doxorubicin Toxicity Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8898919. [PMID: 34035878 PMCID: PMC8116148 DOI: 10.1155/2021/8898919] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 01/13/2021] [Accepted: 04/19/2021] [Indexed: 11/17/2022]
Abstract
Experimental studies have shown the action of green tea in modulating cardiac remodeling. However, the effects of green tea on the cardiac remodeling process induced by doxorubicin (DOX) are not known. Therefore, this study is aimed at evaluating whether green tea extract could attenuate DOX-induced cardiac remodeling, assessed by cardiac morphological and functional changes and associated with the evaluation of different modulators of cardiac remodeling. The animals were divided into four groups: the control group (C), the green tea group (GT), the DOX group (D), and the DOX and green tea group (DGT). Groups C and GT received intraperitoneal sterile saline injections, D and DGT received intraperitoneal injections of DOX, and GT and DGT were fed chow supplemented with green tea extract for 35 days prior to DOX injection. After forty-eight hours, we performed an echocardiogram and euthanasia and collected the materials for analysis. Green tea attenuated DOX-induced cardiotoxicity by increasing cardiac function and decreasing the concentric remodeling. Treatment with DOX increased oxidative stress in the heart, marked by a higher level of lipid hydroperoxide (LH) and lower levels of antioxidant enzymes. Treatment with green tea increased the antioxidant enzymes' activity and decreased the production of LH. Green tea extract increased the expression of Top2-β independent of DOX treatment. The activity of ATP synthase, citrate synthase, and complexes I and II decreased with DOX, without the effects of green tea. Both groups that received DOX presented with a lower ratio of P-akt/T-akt and a higher expression of CD45, TNFα, and intermediate MMP-2, without the effects of green tea. In conclusion, green tea attenuated cardiac remodeling induced by DOX and was associated with increasing the expression of Top2-β and lowering oxidative stress. However, energy metabolism and inflammation probably do not receive the benefits induced by green tea in this model.
Collapse
|
17
|
Yu H, Qiu Y, Yu H, Wang Z, Xu J, Peng Y, Wan X, Wu X, Jin R, Zhou F. Anthracycline Induced Cardiac Disorders in Childhood Acute Lymphoblastic Leukemia: A Single-Centre, Retrospective, Observational Study. Front Pharmacol 2021; 12:598708. [PMID: 33854429 PMCID: PMC8039458 DOI: 10.3389/fphar.2021.598708] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 02/12/2021] [Indexed: 12/04/2022] Open
Abstract
Anthracycline-associated cardiotoxicity is frequently seen in cancer survivors years after treatment, but it is rare in patients on chemotherapy. This study aimed to investigate the clinical characteristics of cardiac disorders in children with acute lymphoblastic leukemia (ALL) during chemotherapy. A retrospective case study was conducted in children with ALL, for whom electrocardiogram (ECG) and echocardiography (Echo) were regularly assessed before each course of chemotherapy. The cardiac disorders were diagnosed according to the Common Terminology Criteria for Adverse Events (CTCAE) Version 5.0. Binary logistic regression analysis was used to identify risk factors associated with cardiac disorders. There were 171 children eligible for the study, and 78 patients (45.61%) were confirmed as having cardiac disorders. The incidence of cardiac disorders was dependent upon the cumulative dose of daunorubicin (DNR) (p = 0.030, OR = 1.553, 95% CI: 1.005–3.108). Four patients (2.34%) presented with palpitation, chest pain, and persistent tachycardia, and they were cured or improved after medical intervention. A total of 74 patients (43.27%) had subclinical cardiac disorders confirmed by ECG or Echo. ECG abnormalities were commonly seen in the induction and continuation treatments, including arrhythmias (26, 15.20%), ST changes (24, 14.04%) and conduction disorders (4, 2.34%). Pericardial effusion (14, 8.19%), left ventricular hypertrophy (11, 6.43%), a widened pulmonary artery (5, 2.92%) and valvular insufficiency (5, 2.92%) suggested by Echo occurred after induction chemotherapy. Therefore, cardiac disorders with clinical manifestations are rare and need early intervention. Subclinical cardiac disorders are common but very hidden in children during ALL chemotherapy. Regular ECG and Echo could help paediatricians to identify and monitor patients with asymptomatic cardiac disorders earlier.
Collapse
Affiliation(s)
- Hui Yu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yining Qiu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hui Yu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhujun Wang
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiawei Xu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yun Peng
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xia Wan
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaoyan Wu
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Runming Jin
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fen Zhou
- Department of Pediatrics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
18
|
Inbody SC, Sinquefield BE, Lewis JP, Horton RE. Biomimetic microsystems for cardiovascular studies. Am J Physiol Cell Physiol 2021; 320:C850-C872. [PMID: 33760660 DOI: 10.1152/ajpcell.00026.2020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Traditional tissue culture platforms have been around for several decades and have enabled key findings in the cardiovascular field. However, these platforms failed to recreate the mechanical and dynamic features found within the body. Organs-on-chips (OOCs) are cellularized microfluidic-based devices that can mimic the basic structure, function, and responses of organs. These systems have been successfully utilized in disease, development, and drug studies. OOCs are designed to recapitulate the mechanical, electrical, chemical, and structural features of the in vivo microenvironment. Here, we review cardiovascular-themed OOC studies, design considerations, and techniques used to generate these cellularized devices. Furthermore, we will highlight the advantages of OOC models over traditional cell culture vessels, discuss implementation challenges, and provide perspectives on the state of the field.
Collapse
Affiliation(s)
- Shelby C Inbody
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Bridgett E Sinquefield
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Joshua P Lewis
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| | - Renita E Horton
- Cardiovascular Tissue Engineering Laboratory, Biomedical Engineering Department, Cullen College of Engineering, University of Houston, Houston, Texas
| |
Collapse
|
19
|
Wang Z, Gao J, Teng H, Peng J. Effects of Doxorubicin on Heme Biosynthesis and Metabolism in Cardiomyocyte. Arq Bras Cardiol 2021; 116:315-322. [PMID: 33656082 PMCID: PMC7909973 DOI: 10.36660/abc.20190437] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Accepted: 12/27/2019] [Indexed: 11/23/2022] Open
Abstract
Fundamento A doxorrubicina está associada à cardiotoxicidade e à morbidade cardíaca tardia. O heme está relacionado ao stress oxidativo celular. Entretanto, sua regulação específica em cardiomiócitos sob os efeitos de doxorrubicina ainda não foi documentada. Objetivo Nosso objetivo é avaliar as alterações de enzimas limitantes de velocidade no caminho metabólico do heme sob o efeito de doxorrubicina. Métodos Cardiomiócitos H9c2 com doxorrubicina em concentrações diferentes (1, 2, 5, 10μM respectivamente). Os testes de PCR em tempo real e Western Blot foram usados para determinar a expressão de proteína e mRNA para quatro enzimas cruciais (ALAS1, ALAS2, HOX-1, e HOX-2) que regulam o metabolismo do heme celular, e os níveis de heme foram detectados por ELISA. Um p<0,01 foi considerado significativo. Resultados Observamos um padrão com alteração dependendo da dose nos níveis de heme nas células H9c2 com o nível mais alto na concentração de 5μM de doxorrubicina, o que ocorreu sincronicamente com o nível mais alto de regulação para cima de ALAS1, bem como as enzimas degenerativas HOX-1 e HOX-2 na expressão de proteína e mRNA. Em contraste, observamos que a ALAS2 foi regulada para baixo gradualmente, inversamente proporcional às concentrações de doxorrubicina. Conclusão O aumento da expressão de ALAS1 pode ter um papel na elevação do nível do heme quando o cardiomiócito H9c2 for exposto à doxorrubicina, e pode ser um alvo terapêutico para a toxicidade miocárdica induzida por doxorrubicina. (Arq Bras Cardiol. 2021; 116(2):315-322)
Collapse
Affiliation(s)
- Zuoyan Wang
- Capital Medical University Affiliated Beijing Shijitan Hospital,Beijing - China
| | - Junyi Gao
- Capital Medical University Affiliated Beijing Shijitan Hospital,Beijing - China
| | - Haobo Teng
- Capital Medical University Affiliated Beijing Shijitan Hospital,Beijing - China
| | - Jianjun Peng
- Capital Medical University Affiliated Beijing Shijitan Hospital,Beijing - China
| |
Collapse
|
20
|
Olorundare OE, Adeneye AA, Akinsola AO, Ajayi AM, Agede OA, Soyemi SS, Mgbehoma AI, Okoye II, Albrecht RM, Ntambi JM, Crooks PA. Therapeutic Potentials of Selected Antihypertensive Agents and Their Fixed-Dose Combinations Against Trastuzumab-Mediated Cardiotoxicity. Front Pharmacol 2021; 11:610331. [PMID: 33897413 PMCID: PMC8058606 DOI: 10.3389/fphar.2020.610331] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 11/23/2020] [Indexed: 01/20/2023] Open
Abstract
Trastuzumab (TZM) is useful in the clinical management of HER2-positive metastatic breast, gastric, and colorectal carcinoma but has been limited by its off-target cardiotoxicity. This study investigates the therapeutic potentials of 0.25 mg/kg/day amlodipine, 0.035 mg/kg/day lisinopril, 5 mg/kg/day valsartan, and their fixed-dose combinations in TZM-intoxicated Wistar rats that were randomly allotted into 10 groups of 6 rats for each group. Group I rats were treated with 10 ml/kg/day sterile water orally and 1 ml/kg/day sterile water intraperitoneally; Groups II, III, and IV rats were orally gavaged with 5 mg/kg/day valsartan and 1 ml/kg/day sterile water intraperitoneally, 0.25 mg/kg/day amlodipine and 1 ml/kg/day sterile water via the intraperitoneal route, 0.035 mg/kg/day lisinopril and 1 ml/kg/day sterile water administered intraperitoneally, respectively. Group V rats were orally treated with 10 ml/kg/day of sterile water prior to intraperitoneal administration of 2.25 mg/kg/day of TZM. Groups VI–VIII rats were equally pretreated with 5 mg/kg/day valsartan, 0.25 mg/kg/day amlodipine, and 0.035 mg/kg/day lisinopril before intraperitoneal 2.25 mg/kg/day TZM treatment, respectively; Groups IX and X rats were orally pretreated with the fixed-dose combinations of 0.25 mg/kg/day amlodipine +0.035 mg/kg/day lisinopril and 5 mg/kg/day valsartan +0.035 mg/kg/day lisinopril, respectively, before TZM treatment. Cardiac injury and tissue oxidative stress markers, complete lipids profile, histopathological, and immunohistochemical assays were the evaluating endpoints. Results showed that repeated TZM treatments caused profound increases in the serum TG and VLDL-c levels, serum cTnI and LDH levels, and cardiac tissue caspase-3 and -9 levels but decreased BCL-2 expression. TZM also profoundly attenuated CAT, SOD, GST and GPx activities, and increased MDA levels in the treated tissues. In addition, TZM cardiotoxicity was characterized by marked vascular and cardiomyocyte congestion and coronary artery microthrombi formation. However, the altered biochemical, histopathological, and immunohistochemical changes were reversed with amlodipine, lisinopril, valsartan, and fixed-dose combinations, although fixed-dose valsartan/lisinopril combination was further associated with hyperlipidemia and increased AI and CRI values and coronary artery cartilaginous metaplasia. Thus, the promising therapeutic potentials of amlodipine, lisinopril, valsartan and their fixed-dose combinations in the management of TZM cardiotoxicity, majorly mediated via antiapoptotic and oxidative stress inhibition mechanisms were unveiled through this study.
Collapse
Affiliation(s)
- Olufunke Esan Olorundare
- Department of Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Adejuwon Adewale Adeneye
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, Ikeja, Nigeria
| | - Akinyele Olubiyi Akinsola
- Department of Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Abayomi Mayowa Ajayi
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Nigeria
| | - Olalekan Ayodele Agede
- Department of Pharmacology and Therapeutics, Faculty of Basic Clinical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Sunday Sokunle Soyemi
- Department of Pathology and Forensic Medicine, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, Ikeja, Nigeria
| | - Alban Ikenna Mgbehoma
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, Ikeja, Nigeria
| | - Ikechukwu Innocent Okoye
- Department of Oral Pathology and Medicine, Faculty of Dentistry, Lagos State University College of Medicine, Ikeja, Nigeria
| | - Ralph M Albrecht
- Department of Animal Sciences, University of Wisconsin, Madison, WI, United States
| | - James Mukasa Ntambi
- Department of Nutritional Sciences, College of Agricultural and Life Sciences, University of Wisconsin, Madison, Madison, WI, United States
| | - Peter Anthony Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
21
|
Kihara M, Kaiya H, Hirai Y, Katayama H, Terao A, Nishikawa M. Salmon acyl-ghrelin increases food intake and reduces doxorubicin-induced myocardial apoptosis in rats, likely by anti-oxidative activity. Peptides 2021; 137:170471. [PMID: 33340558 DOI: 10.1016/j.peptides.2020.170471] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 10/26/2020] [Accepted: 11/28/2020] [Indexed: 12/16/2022]
Abstract
We had reported that orally administered ghrelin-containing salmon stomach extract prevents doxorubicin (DOX)-induced cardiotoxicity. In this study, we investigated the binding affinity of salmon ghrelin to rat ghrelin receptor and the cardioprotective effects of subcutaneous (sc) injected synthetic salmon ghrelin in rats with DOX-induced acute heart failure in order to clarify the potential efficacy of salmon ghrelin. Intracellular calcium mobilization assay was performed on rat GHS-R1a-expressing CHO cells to reveal ghrelin activity. Rats were divided into five groups; the normal control (I), and toxic control (II) groups were given saline (sc, twice daily), and the salmon acyl-ghrelin (sAG) (III), salmon unacylated-ghrelin (sUAG) (IV), and rat acyl-ghrelin (rAG) (V) groups were given corresponding synthetic ghrelins (sc, twice daily), respectively. After seven days of treatment, DOX (20 mg/kg BW) or saline was administered to the corresponding groups by intraperitoneal injection. The toxic control group was the negative control group for the DOX-induced cardiotoxicity groups. While sAG displayed similar affinity to rAG upon application to GHS-R1a-expressing cells, and also decreased DOX-induced apoptosis and increased food intake, sUAG did not. Both sAG and rAG improved DOX-induced deterioration, showing anti-oxidative activity. The anti-oxidative activity of sAG might contribute to the protective effects on cardiomyocytes. The results also suggest that, similar to rAG, sAG is a potent protectant against DOX-induced cardiotoxicity and a potential functional component in orally administered ghrelin-containing salmon stomach extract, which prevented DOX-induced cardiotoxicity in our previous study.
Collapse
Affiliation(s)
- Minoru Kihara
- Department of Marine Biology and Sciences, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan.
| | - Hiroyuki Kaiya
- Department of Biochemistry, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe-shinmachi, Suita, 564-8565, Japan
| | - Yumi Hirai
- Department of Marine Biology and Sciences, School of Biological Sciences, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan
| | - Hidekazu Katayama
- Department of Applied Biochemistry, School of Engineering, Tokai University, 4-1-1 Kitakaname, Hiratsuka, Kanagawa 259-1292, Japan
| | - Akira Terao
- Liberal Arts Education Center, Sapporo Campus, Tokai University, 5-1-1-1 Minamisawa, Minami-ku, Sapporo, 005-8601, Japan
| | - Masazumi Nishikawa
- Department of Food Management, School of Food, Agricultural and Environmental Sciences, Miyagi University, 2-2-1 Hatadate, Taihaku-ku, Sendai, 982-0215, Japan
| |
Collapse
|
22
|
Gatto M, Mota GAF. Influence of Doxorubicin Treatment on Heme Metabolism in Cardiomyoblasts: An In Vitro Study. Arq Bras Cardiol 2021; 116:323-324. [PMID: 33656083 PMCID: PMC7909966 DOI: 10.36660/abc.20200662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Mariana Gatto
- Departamento de Clínica MédicaFaculdade de Medicina de BotucatuUniversidade Estadual PaulistaBotucatuSPBrasilDepartamento de Clínica Médica, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista - Unesp, Botucatu, SP - Brasil
| | - Gustavo Augusto Ferreira Mota
- Departamento de Clínica MédicaFaculdade de Medicina de BotucatuUniversidade Estadual PaulistaBotucatuSPBrasilDepartamento de Clínica Médica, Faculdade de Medicina de Botucatu, Universidade Estadual Paulista - Unesp, Botucatu, SP - Brasil
| |
Collapse
|
23
|
The Metabolic Role of GRK2 in Insulin Resistance and Associated Conditions. Cells 2021; 10:cells10010167. [PMID: 33467677 PMCID: PMC7830135 DOI: 10.3390/cells10010167] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 02/07/2023] Open
Abstract
Insulin resistance (IRES) is a pathophysiological condition characterized by the reduced response to insulin of several tissues, including myocardial and skeletal muscle. IRES is associated with obesity, glucose intolerance, dyslipidemia, and hypertension, evolves toward type 2 diabetes, and increases the risk of developing cardiovascular diseases. Several studies designed to explore the mechanisms involved in IRES allowed the identification of a multitude of potential molecular targets. Among the most promising, G Protein Coupled Receptor Kinase type 2 (GRK2) appears to be a suitable one given its functional implications in many cellular processes. In this review, we will discuss the metabolic role of GRK2 in those conditions that are characterized by insulin resistance (diabetes, hypertension, heart failure), and the potentiality of its inhibition as a therapeutic strategy to revert both insulin resistance and its associated phenotypes.
Collapse
|
24
|
Dame K, Ribeiro AJ. Microengineered systems with iPSC-derived cardiac and hepatic cells to evaluate drug adverse effects. Exp Biol Med (Maywood) 2020; 246:317-331. [PMID: 32938227 PMCID: PMC7859673 DOI: 10.1177/1535370220959598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic and cardiac drug adverse effects are among the leading causes of attrition in drug development programs, in part due to predictive failures of current animal or in vitro models. Hepatocytes and cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) hold promise for predicting clinical drug effects, given their human-specific properties and their ability to harbor genetically determined characteristics that underlie inter-individual variations in drug response. Currently, the fetal-like properties and heterogeneity of hepatocytes and cardiomyocytes differentiated from iPSCs make them physiologically different from their counterparts isolated from primary tissues and limit their use for predicting clinical drug effects. To address this hurdle, there have been ongoing advances in differentiation and maturation protocols to improve the quality and use of iPSC-differentiated lineages. Among these are in vitro hepatic and cardiac cellular microsystems that can further enhance the physiology of cultured cells, can be used to better predict drug adverse effects, and investigate drug metabolism, pharmacokinetics, and pharmacodynamics to facilitate successful drug development. In this article, we discuss how cellular microsystems can establish microenvironments for these applications and propose how they could be used for potentially controlling the differentiation of hepatocytes or cardiomyocytes. The physiological relevance of cells is enhanced in cellular microsystems by simulating properties of tissue microenvironments, such as structural dimensionality, media flow, microfluidic control of media composition, and co-cultures with interacting cell types. Recent studies demonstrated that these properties also affect iPSC differentiations and we further elaborate on how they could control differentiation efficiency in microengineered devices. In summary, we describe recent advances in the field of cellular microsystems that can control the differentiation and maturation of hepatocytes and cardiomyocytes for drug evaluation. We also propose how future research with iPSCs within engineered microenvironments could enable their differentiation for scalable evaluations of drug effects.
Collapse
Affiliation(s)
- Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alexandre Js Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
25
|
Lopes GM, Grudzinski PB, Beyer Nardi N, Leguisamo NM. Cell Therapy Improves Cardiac Function in Anthracycline-Induced Cardiomyopathy Preclinical Models: A Systematic Review and Meta-Analysis. Stem Cells Dev 2020; 29:1247-1265. [PMID: 32741268 DOI: 10.1089/scd.2020.0044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Although anthracycline (ANT)-based treatment strongly contributes to cancer survivorship, the use of these agents is limited by the risk of cardiotoxicity. For those patients who evolve to heart failure, myocardial regenerative approaches are of particular interest, and a growing body of preclinical studies has been investigating the use of cell therapy for ANT-induced cardiomyopathy (AIC). However, since animal models and modalities of cell therapy are highly heterogeneous between studies, the efficacy of cell therapy for AIC is not clear. Thus, we conducted a systematic review and meta-analysis of experimental studies reporting the use of cell therapy with mesenchymal stromal cells (MSC) or bone marrow mononuclear cells (BMMNC) in animal models of AIC with regard to global cardiac function. The Medline, EMBASE, and Web of Science databases were searched from inception to November 2019. Two reviewers independently extracted data on study quality and the results of left ventricular ejection fraction (LVEF) and fractional shortening (FS) obtained by echocardiography. The quality of outcomes was assessed using the Cochrane, Collaborative Approach to Meta-Analysis and Review of Animal Data from Experimental Studies (CAMARADES), and SYRCLE bias risk tools. Pooled random-effects modeling was used to calculate pooled mean differences (MD) and 95% confidence intervals (CIs). Twenty-two studies comprising 381 small animals (rabbits and rodents) were included. A pooled meta-analysis of all treatments showed that cell therapy increased LVEF by 9.87% (95% CI 7.25-12.50, P < 0.00001) and FS by 7.80% (95% CI 5.68-9.92, P < 0.00001) in small animals with AIC. Cell therapy with MSC/BMMNC is effective to mitigate the deleterious effects of ANT on cardiac function in preclinical models. Nevertheless, due to the small number of studies and considerable heterogeneity, future translational studies must be designed to diminish between-study discrepancies and increase similarity to the clinical landscape.
Collapse
Affiliation(s)
- Gabriela Maciel Lopes
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil.,Graduate Program in Health Sciences (Cardiology), University Foundation of Cardiology, Porto Alegre, Brazil
| | - Patrícia Bencke Grudzinski
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil
| | - Nance Beyer Nardi
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil.,Graduate Program in Health Sciences (Cardiology), University Foundation of Cardiology, Porto Alegre, Brazil
| | - Natalia Motta Leguisamo
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology (IC/FUC), Porto Alegre, Brazil.,Graduate Program in Health Sciences (Cardiology), University Foundation of Cardiology, Porto Alegre, Brazil
| |
Collapse
|
26
|
Borovac JA, D'Amario D, Bozic J, Glavas D. Sympathetic nervous system activation and heart failure: Current state of evidence and the pathophysiology in the light of novel biomarkers. World J Cardiol 2020; 12:373-408. [PMID: 32879702 PMCID: PMC7439452 DOI: 10.4330/wjc.v12.i8.373] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/19/2020] [Accepted: 07/19/2020] [Indexed: 02/06/2023] Open
Abstract
Heart failure (HF) is a complex clinical syndrome characterized by the activation of at least several neurohumoral pathways that have a common role in maintaining cardiac output and adequate perfusion pressure of target organs and tissues. The sympathetic nervous system (SNS) is upregulated in HF as evident in dysfunctional baroreceptor and chemoreceptor reflexes, circulating and neuronal catecholamine spillover, attenuated parasympathetic response, and augmented sympathetic outflow to the heart, kidneys and skeletal muscles. When these sympathoexcitatory effects on the cardiovascular system are sustained chronically they initiate the vicious circle of HF progression and become associated with cardiomyocyte apoptosis, maladaptive ventricular and vascular remodeling, arrhythmogenesis, and poor prognosis in patients with HF. These detrimental effects of SNS activity on outcomes in HF warrant adequate diagnostic and treatment modalities. Therefore, this review summarizes basic physiological concepts about the interaction of SNS with the cardiovascular system and highlights key pathophysiological mechanisms of SNS derangement in HF. Finally, special emphasis in this review is placed on the integrative and up-to-date overview of diagnostic modalities such as SNS imaging methods and novel laboratory biomarkers that could aid in the assessment of the degree of SNS activation and provide reliable prognostic information among patients with HF.
Collapse
Affiliation(s)
- Josip Anđelo Borovac
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
- Working Group on Heart Failure of Croatian Cardiac Society, Zagreb 10000, Croatia
| | - Domenico D'Amario
- Department of Cardiovascular and Thoracic Sciences, IRCCS Fondazione Policlinico A. Gemelli, Universita Cattolica Sacro Cuore, Rome 00168, Italy
| | - Josko Bozic
- Department of Pathophysiology, University of Split School of Medicine, Split 21000, Croatia
| | - Duska Glavas
- Working Group on Heart Failure of Croatian Cardiac Society, Zagreb 10000, Croatia
- Clinic for Cardiovascular Diseases, University Hospital of Split, Split 21000, Croatia
| |
Collapse
|
27
|
Selective protection of human cardiomyocytes from anthracycline cardiotoxicity by small molecule inhibitors of MAP4K4. Sci Rep 2020; 10:12060. [PMID: 32694738 PMCID: PMC7374628 DOI: 10.1038/s41598-020-68907-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 06/25/2020] [Indexed: 12/11/2022] Open
Abstract
Given the poor track record to date of animal models for creating cardioprotective drugs, human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) have been proposed as a therapeutically relevant human platform to guide target validation and cardiac drug development. Mitogen-Activated Protein Kinase Kinase Kinase Kinase-4 (MAP4K4) is an “upstream” member of the MAPK superfamily that is implicated in human cardiac muscle cell death from oxidative stress, based on gene silencing and pharmacological inhibition in hPSC-CMs. A further role for MAP4K4 was proposed in heart muscle cell death triggered by cardiotoxic anti-cancer drugs, given its reported activation in failing human hearts with doxorubicin (DOX) cardiomyopathy, and its activation acutely by DOX in cultured cardiomyocytes. Here, we report successful protection from DOX in two independent hPSC-CM lines, using two potent, highly selective MAP4K4 inhibitors. The MAP4K4 inhibitors enhanced viability and reduced apoptosis at otherwise lethal concentrations of DOX, and preserved cardiomyocyte function, as measured by spontaneous calcium transients, at sub-maximal ones. Notably, in contrast, no intereference was seen in tumor cell killing, caspase activation, or mitochondrial membrane dissipation by DOX, in human cancer cell lines. Thus, MAP4K4 is a plausible, tractable, selective therapeutic target in DOX-induced human heart muscle cell death.
Collapse
|
28
|
Quagliariello V, Vecchione R, De Capua A, Lagreca E, Iaffaioli RV, Botti G, Netti PA, Maurea N. Nano-Encapsulation of Coenzyme Q10 in Secondary and Tertiary Nano-Emulsions for Enhanced Cardioprotection and Hepatoprotection in Human Cardiomyocytes and Hepatocytes During Exposure to Anthracyclines and Trastuzumab. Int J Nanomedicine 2020; 15:4859-4876. [PMID: 32764923 PMCID: PMC7359894 DOI: 10.2147/ijn.s245170] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Accepted: 04/18/2020] [Indexed: 12/19/2022] Open
Abstract
INTRODUCTION CoenzymeQ10 (CoQ10) is a well-known antioxidant and anti-inflammatory agent with cardioprotective properties. However, clinical trials based on its oral administration have failed to provide significant effect on cardiac functionality. The main limitation of CoQ10 is based on its very low oral bioavailability and instability that limit dramatically its effects as a cardioprotective agent. Herein, we loaded CoQ10 in high bioavailable nano-emulsions (NEs) coated with chitosan or chitosan and hyaluronic acid in order to improve its performance. METHODS We tested cardioprotective and hepatoprotective effects of CoQ10-loaded nano-carriers against Doxorubicin and Trastuzumab toxicities in cardiomyocytes and liver cells through analysis of cell viability, lipid peroxidation, expression of leukotrienes, p65/NF-kB and pro-inflammatory cytokines involved in anticancer-induced cardio and hepatotoxicity. RESULTS Nano-carriers showed high stability and loading ability and increased cell viability both in hepatocytes and cardiomyocytes during anticancer treatments. We observed that these effects are mediated by the inhibition of lipid peroxidation and reduction of the inflammation. CoQ10-loaded nano-emulsions showed also strong anti-inflammatory effects reducing leukotriene B4 and p65/NF-κB expression and Interleukin 1β and 6 production during anticancer treatments. DISCUSSION Anthracyclines and Human epidermal growth factor receptor (HER2) inhibitors have shown significant anticancer effects in clinical practice but their use is characterized by cardiotoxicity and hepatotoxicity. Nano-carriers loaded with CoQ10 showed cardio and hepatoprotective properties mediated by reduction of oxidative damages and pro-inflammatory mediators. These results set the stage for preclinical studies of cardio and hepatoprotection in HER2+ breast cancer-bearing mice treated with Doxorubicin and Trastuzumab.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, Napoli, Italia
| | - Raffaele Vecchione
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | - Alberta De Capua
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | - Elena Lagreca
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | | | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, Napoli, Italia
| | - Paolo A Netti
- Center for Advanced Biomaterial for Health Care (CABHC), Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, Naples, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori- IRCCS- Fondazione G. Pascale, Napoli, Italia
| |
Collapse
|
29
|
Jiang L, Ping L, Yan H, Yang X, He Q, Xu Z, Luo P. Cardiovascular toxicity induced by anti-VEGF/VEGFR agents: a special focus on definitions, diagnoses, mechanisms and management. Expert Opin Drug Metab Toxicol 2020; 16:823-835. [PMID: 32597258 DOI: 10.1080/17425255.2020.1787986] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Vascular endothelial growth factor (VEGF) is a key target in cancer therapy. However, cardiovascular safety has been one of the most challenging aspects of anti-VEGF/VEGF receptor (VEGFR) agent development and therapy. While accurate diagnostic modalities for assessment of cardiac function have been developed over the past few decades, a lack of an optimal definition and precise mechanism still places a significant limit on the effective management of cardiovascular toxicity. AREAS COVERED Here, we report the cardiovascular toxicity profile associated with anti-VEGF/VEGFR agents and summarize the clinical diagnoses as well as management that are already performed in clinical practice or are currently being investigated. Furthermore, the review discusses the potential molecular toxicological mechanisms, which may provide strategies to prevent toxicity and drive drug discovery. EXPERT OPINION Cardiovascular toxicity associated with anti-VEGF/VEGFR agents has been a substantial risk for cancer treatment. To improve its management, the development of guidelines for prevention, monitoring and treatment of cardiovascular toxicity has become a hot topic. The summary of cardiovascular toxicity profile, mechanisms and management given in this review is not only significant for the optimal use of existing anti-VEGF/VEGFR agents to protect patients predisposed to cardiovascular toxicity but is also beneficial for drug development.
Collapse
Affiliation(s)
- Liyu Jiang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Li Ping
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Hao Yan
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Zhifei Xu
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| | - Peihua Luo
- Center for Drug Safety Evaluation and Research of Zhejiang University, College of Pharmaceutical Sciences, Zhejiang University , Hangzhou, Zhejiang,China
| |
Collapse
|
30
|
Blair CA, Pruitt BL. Mechanobiology Assays with Applications in Cardiomyocyte Biology and Cardiotoxicity. Adv Healthc Mater 2020; 9:e1901656. [PMID: 32270928 PMCID: PMC7480481 DOI: 10.1002/adhm.201901656] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/31/2020] [Accepted: 02/03/2020] [Indexed: 12/19/2022]
Abstract
Cardiomyocytes are the motor units that drive the contraction and relaxation of the heart. Traditionally, testing of drugs for cardiotoxic effects has relied on primary cardiomyocytes from animal models and focused on short-term, electrophysiological, and arrhythmogenic effects. However, primary cardiomyocytes present challenges arising from their limited viability in culture, and tissue from animal models suffers from a mismatch in their physiology to that of human heart muscle. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) can address these challenges. They also offer the potential to study not only electrophysiological effects but also changes in cardiomyocyte contractile and mechanical function in response to cardiotoxic drugs. With growing recognition of the long-term cardiotoxic effects of some drugs on subcellular structure and function, there is increasing interest in using hiPSC-CMs for in vitro cardiotoxicity studies. This review provides a brief overview of techniques that can be used to quantify changes in the active force that cardiomyocytes generate and variations in their inherent stiffness in response to cardiotoxic drugs. It concludes by discussing the application of these tools in understanding how cardiotoxic drugs directly impact the mechanobiology of cardiomyocytes and how cardiomyocytes sense and respond to mechanical load at the cellular level.
Collapse
Affiliation(s)
- Cheavar A. Blair
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Beth L. Pruitt
- Department of mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, CA, USA
- Molecular, Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| |
Collapse
|
31
|
Piper-Vallillo AJ, Costa DB, Sabe MA, Asnani A. Heart Failure Associated With the Epidermal Growth Factor Receptor Inhibitor Osimertinib. JACC CardioOncol 2020; 2:119-122. [PMID: 34396216 PMCID: PMC8352313 DOI: 10.1016/j.jaccao.2020.01.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Revised: 01/07/2020] [Accepted: 01/13/2020] [Indexed: 12/28/2022] Open
Affiliation(s)
- Andrew J. Piper-Vallillo
- Department of Medicine, Division of Medical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel B. Costa
- Department of Medicine, Division of Medical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Marwa A. Sabe
- Advanced Heart Failure and Mechanical Circulatory Support Program, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Aarti Asnani
- Cardio-Oncology Program and CardioVascular Institute, Beth Israel Deaconess Medical Institute, Beth Israel Deaconess Medical Center/Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Duraes AR, Filho CRH, de Souza Lima Bitar Y, Neto MG. Heart Failure and Comorbidities—Part 1. CURRENT EMERGENCY AND HOSPITAL MEDICINE REPORTS 2020. [DOI: 10.1007/s40138-020-00210-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
33
|
Akentieva NP, Sanina NA, Gizatullin AR, Shkondina NI, Prikhodchenko TR, Shram SI, Zhelev N, Aldoshin SM. Cytoprotective Effects of Dinitrosyl Iron Complexes on Viability of Human Fibroblasts and Cardiomyocytes. Front Pharmacol 2019; 10:1277. [PMID: 31780929 PMCID: PMC6859909 DOI: 10.3389/fphar.2019.01277] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 10/07/2019] [Indexed: 12/29/2022] Open
Abstract
Nitric oxide (NO) is an important signaling molecule that plays a key role in maintaining vascular homeostasis. Dinitrosyl iron complexes (DNICs) generating NO are widely used to treat cardiovascular diseases. However, the involvement of DNICs in the metabolic processes of the cell, their protective properties in doxorubicin-induced toxicity remain to be clarified. Here, we found that novel class of mononuclear DNICs with functional sulfur-containing ligands enhanced the cell viability of human lung fibroblasts and rat cardiomyocytes. Moreover, DNICs demonstrated remarkable protection against doxorubicin-induced toxicity in fibroblasts and in rat cardiomyocytes (H9c2 cells). Data revealed that the DNICs compounds modulate the mitochondria function by decreasing the mitochondrial membrane potential (ΔΨm). Results of flow cytometry showed that DNICs were not affected the proliferation, growth of fibroblasts. In addition, this study showed that DNICs did not affect glutathione levels and the formation of reactive oxygen species in cells. Moreover, results indicated that DNICs maintained the ATP equilibrium in cells. Taken together, these findings show that DNICs have protective properties in vitro. It was further suggested that DNICs may be uncouplers of oxidative phosphorylation in mitochondria and protective mechanism is mainly provided by the leakage of excess charge through the mitochondrial membrane. It is assumed that the DNICs have the therapeutic potential for treating cardiovascular diseases and for decreasing of chemotherapy-induced cardiotoxicity in cancer survivors.
Collapse
Affiliation(s)
- Natalia Pavlovna Akentieva
- Laboratory Biochemical and Cellular Studies, Department of Kinetics of Chemical and Biological Processes, Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Russia
- Laboratory of Toxicology and Experimental Chemotherapy, Moscow State Regional University, Moscow, Russia
- Faculty of Medicine, Karabük University, Karabük, Turkey
| | - Natalia Alekseevna Sanina
- Laboratory of Structural Chemistry, Department of Structure of Matter, Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Russia
- Faculty of fundamental physical and chemical engineering, Lomonosov Moscow State University, Moscow, Russia
| | - Artur Rasimovich Gizatullin
- Laboratory Biochemical and Cellular Studies, Department of Kinetics of Chemical and Biological Processes, Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Russia
| | - Natalia Ivanovna Shkondina
- Laboratory Biochemical and Cellular Studies, Department of Kinetics of Chemical and Biological Processes, Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Russia
| | - Tatyana Romanovna Prikhodchenko
- Laboratory Biochemical and Cellular Studies, Department of Kinetics of Chemical and Biological Processes, Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Russia
| | - Stanislav Ivanovich Shram
- Neuropharmacology Sector, Institute of Molecular Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Nikolai Zhelev
- School of Medicine, University of Dundee, Dundee, United Kingdom
- Medical University Plovdiv, Plovdiv, Bulgaria
| | - Sergei Michailovich Aldoshin
- Laboratory of Structural Chemistry, Department of Structure of Matter, Institute of Problems of Chemical Physics, Russian Academy of Sciences, Chernogolovka, Russia
- Faculty of fundamental physical and chemical engineering, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|